Abstract A08: Combination with a novel STING agonist significantly improves efficacy of anti-PD1 therapy in mouse syngeneic tumor models

Samanthi A. Perera, E. KopinjaJohnny, Yanhong Ma, J. Laskey, K. Chakravarthy, L. Cui, Yiping Chen, J. Presland, Sharad K Sharma, Shuxia Zhao, J. Piesvaux, E. Minnihan, Heidi Ferguson, Hyun Woo, Ian Knemeyer, I. Kariv, A. Tse, S. Cemerski, J. Cumming, B. Trotter, B. Pan, G. Addona, B. Long
{"title":"Abstract A08: Combination with a novel STING agonist significantly improves efficacy of anti-PD1 therapy in mouse syngeneic tumor models","authors":"Samanthi A. Perera, E. KopinjaJohnny, Yanhong Ma, J. Laskey, K. Chakravarthy, L. Cui, Yiping Chen, J. Presland, Sharad K Sharma, Shuxia Zhao, J. Piesvaux, E. Minnihan, Heidi Ferguson, Hyun Woo, Ian Knemeyer, I. Kariv, A. Tse, S. Cemerski, J. Cumming, B. Trotter, B. Pan, G. Addona, B. Long","doi":"10.1158/2326-6074.TUMIMM17-A08","DOIUrl":null,"url":null,"abstract":"Activated STING (STimulator of INterferon Genes) bound to its natural ligand 2,3-cGAMP (cyclic guanosine-adenosine monophosphate), initiates type I interferon (IFN) and pro-inflammatory cytokine production. IFN upregulation is essential to promote antigen-specific CD8+ T-cell priming and leads to potent anti-tumor activity. To exploit this mechanism we synthesized a new STING agonist, MSA-1, that potently activates both mouse and human STING. Intratumoral (IT) administration of MSA-1 to MC38 syngeneic tumor-bearing mice increased tumor and plasma cytokine levels and was effective at driving complete responses (CRs) in 100% of the animals. Most surviving animals developed tumor-specific adaptive immune memory as demonstrated by robust protection against re-challenge with the same tumor type. Mechanistic studies in immune-deficient mice suggested that the initial antitumor activity is in part due to cytokine-driven cytotoxicity and/or other innate immune mechanisms, which may have contributed to some animals not developing an adaptive immune memory. Importantly, MSA-1 caused long-term tumor regressions or CRs in CT26 and B16-F10 tumor models, both of which are intrinsically resistant to single-agent therapy with a fully murinized anti-mouse PD-1 antibody (muDX400). The antitumor immune response in these models was further enhanced when treating the animals with MSA-1 in combination with muDX400. This combination restored T-cell responses in both blood and tumors of the treated mice and provided long-lived immunologic memory in a majority of the animals. Taken together, these data strongly support the development of STING agonists in combination with Keytruda (humanized anti-PD-1 antibody) for patients with tumors that are partially responsive or nonresponsive to single agent anti-PD-1 therapy. Citation Format: Samanthi A. Perera, Johnny E. Kopinja, Yanhong Ma, Jason Laskey, Kalyan Chakravarthy, Long Cui, Yiping Chen, Jeremy Presland, Sharad Sharma, Shuxia Zhao, Jennifer Piesvaux, Ellen C. Minnihan, Heidi Ferguson, Hyun Chong Woo, Ian Knemeyer, Ilona Kariv, Archie Tse, Saso Cemerski, Jared Cumming, B. Wesley Trotter, Bo-Sheng Pan, George H. Addona, Brian J. Long. Combination with a novel STING agonist significantly improves efficacy of anti-PD1 therapy in mouse syngeneic tumor models [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A08.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Checkpoints and Immunomodulation","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1158/2326-6074.TUMIMM17-A08","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Activated STING (STimulator of INterferon Genes) bound to its natural ligand 2,3-cGAMP (cyclic guanosine-adenosine monophosphate), initiates type I interferon (IFN) and pro-inflammatory cytokine production. IFN upregulation is essential to promote antigen-specific CD8+ T-cell priming and leads to potent anti-tumor activity. To exploit this mechanism we synthesized a new STING agonist, MSA-1, that potently activates both mouse and human STING. Intratumoral (IT) administration of MSA-1 to MC38 syngeneic tumor-bearing mice increased tumor and plasma cytokine levels and was effective at driving complete responses (CRs) in 100% of the animals. Most surviving animals developed tumor-specific adaptive immune memory as demonstrated by robust protection against re-challenge with the same tumor type. Mechanistic studies in immune-deficient mice suggested that the initial antitumor activity is in part due to cytokine-driven cytotoxicity and/or other innate immune mechanisms, which may have contributed to some animals not developing an adaptive immune memory. Importantly, MSA-1 caused long-term tumor regressions or CRs in CT26 and B16-F10 tumor models, both of which are intrinsically resistant to single-agent therapy with a fully murinized anti-mouse PD-1 antibody (muDX400). The antitumor immune response in these models was further enhanced when treating the animals with MSA-1 in combination with muDX400. This combination restored T-cell responses in both blood and tumors of the treated mice and provided long-lived immunologic memory in a majority of the animals. Taken together, these data strongly support the development of STING agonists in combination with Keytruda (humanized anti-PD-1 antibody) for patients with tumors that are partially responsive or nonresponsive to single agent anti-PD-1 therapy. Citation Format: Samanthi A. Perera, Johnny E. Kopinja, Yanhong Ma, Jason Laskey, Kalyan Chakravarthy, Long Cui, Yiping Chen, Jeremy Presland, Sharad Sharma, Shuxia Zhao, Jennifer Piesvaux, Ellen C. Minnihan, Heidi Ferguson, Hyun Chong Woo, Ian Knemeyer, Ilona Kariv, Archie Tse, Saso Cemerski, Jared Cumming, B. Wesley Trotter, Bo-Sheng Pan, George H. Addona, Brian J. Long. Combination with a novel STING agonist significantly improves efficacy of anti-PD1 therapy in mouse syngeneic tumor models [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A08.
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
摘要:联合一种新型STING激动剂可显著提高小鼠同基因肿瘤模型抗pd1治疗的疗效
被激活的STING(干扰素基因刺激因子)与其天然配体2,3- cgamp(环鸟苷-单磷酸腺苷)结合,启动I型干扰素(IFN)和促炎细胞因子的产生。IFN的上调对于促进抗原特异性CD8+ t细胞的启动和导致有效的抗肿瘤活性是必不可少的。为了利用这一机制,我们合成了一种新的STING激动剂MSA-1,它能有效激活小鼠和人的STING。肿瘤内给药MSA-1增加MC38同基因荷瘤小鼠的肿瘤和血浆细胞因子水平,并在100%的动物中有效地驱动完全反应(cr)。大多数存活的动物产生了肿瘤特异性适应性免疫记忆,这证明了对相同肿瘤类型的再次攻击的强大保护。免疫缺陷小鼠的机制研究表明,最初的抗肿瘤活性部分是由于细胞因子驱动的细胞毒性和/或其他先天免疫机制,这可能导致一些动物没有形成适应性免疫记忆。重要的是,MSA-1在CT26和B16-F10肿瘤模型中引起长期肿瘤消退或cr,这两种肿瘤模型本质上对完全鼠化的抗小鼠PD-1抗体(muDX400)单药治疗具有耐药性。当MSA-1与muDX400联合治疗动物时,这些模型的抗肿瘤免疫应答进一步增强。这种组合恢复了治疗小鼠血液和肿瘤中的t细胞反应,并为大多数动物提供了长期的免疫记忆。综上所述,这些数据有力地支持了STING激动剂与Keytruda(人源抗pd -1抗体)联合用于对单药抗pd -1治疗部分反应或无反应的肿瘤患者的开发。引文格式:Samanthi A. Perera, Johnny E. Kopinja, Yanhong Ma, Jason Laskey, Kalyan Chakravarthy, Long Cui, Yiping Chen, Jeremy Presland, Sharad Sharma, Shuxia Zhao, Jennifer Piesvaux, Ellen C. Minnihan, Heidi Ferguson, Hyun Chong Woo, Ian Knemeyer, Ilona Kariv, Archie Tse, Saso Cemerski, Jared Cumming, B. Wesley Trotter, Pan Bo-Sheng, George H. adona, Brian J. Long。联合一种新型STING激动剂可显著提高小鼠同基因肿瘤模型抗pd1治疗的疗效[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫杂志,2018;6(9增刊):摘要nr A08。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Abstract A21: Activation of 4-1BB on liver myeloid cells triggers hepatitis via an interleukin-27 dependent pathway Abstract A11: Ipilimumab protects T cells from the antiproliferative effects of dexamethasone Abstract A29: Multimeric anti-human OX40 induces robust immune responses Abstract A33: Tumor derived T cell clones for evaluation of check point inhibitor therapeutics Abstract A40: Prevalence of TIGIT expression in normal tissues, inflammation, and cancer
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1